The Add-On Effect of Lactobacillus plantarum PS128 in Patients With Parkinson's Disease: A Pilot Study

Chin-Song Lu, Hsiu-Chen Chang, Yi-Hsin Weng, Chiung-Chu Chen, Yi-Shan Kuo, Ying-Chieh Tsai, Chin-Song Lu, Hsiu-Chen Chang, Yi-Hsin Weng, Chiung-Chu Chen, Yi-Shan Kuo, Ying-Chieh Tsai

Abstract

Background: Lactobacillus plantarum PS128 (PS128) is a specific probiotic, known as a psychobiotic, which has been demonstrated to alleviate motor deficits and inhibit neurodegenerative processes in Parkinson's disease (PD)-model mice. We hypothesize that it may also be beneficial to patients with PD based on the possible mechanism via the microbiome-gut-brain axis. Methods: This is an open-label, single-arm, baseline-controlled trial. The eligible participants were scheduled to take 60 billion colony-forming units of PS128 once per night for 12 weeks. Clinical assessments were conducted using the Unified Parkinson's Disease Rating Scale (UPDRS), modified Hoehn and Yahr scale, and change in patient "ON-OFF" diary recording as primary outcome measures. The non-motor symptoms questionnaire, Beck depression inventory-II, patient assessment of constipation symptom, 39-item Parkinson's Disease Questionnaire (PDQ-39), and Patient Global Impression of Change (PGI-C) were assessed as secondary outcome measures. Results: Twenty-five eligible patients (32% women) completed the study. The mean age was 61.84 ± 5.74 years (range, 52-72), mean disease duration was 10.12 ± 2.3 years (range, 5-14), and levodopa equivalent daily dosage was 1063.4 ± 209.5 mg/daily (range, 675-1,560). All patients remained on the same dosage of anti-parkinsonian and other drugs throughout the study. After 12 weeks of PS128 supplementation, the UPDRS motor scores improved significantly in both the OFF and ON states (p = 0.004 and p = 0.007, respectively). In addition, PS128 intervention significantly improved the duration of the ON period and OFF period as well as PDQ-39 values. However, no obvious effect of PS128 on non-motor symptoms of patients with PD was observed. Notably, the PGI-C scores improved in 17 patients (68%). PS128 intervention was also found to significantly reduce plasma myeloperoxidase and urine creatinine levels. Conclusion: The present study demonstrated that PS128 supplementation for 12 weeks with constant anti-parkinsonian medication improved the UPDRS motor score and quality of life of PD patients. We suggest that PS128 could serve as a therapeutic adjuvant for the treatment of PD. In the future, placebo-controlled studies are needed to further support the efficacy of PS128 supplementation. Clinical Trial Registration: https://ichgcp.net/clinical-trials-registry/NCT04389762" title="See in ClinicalTrials.gov">NCT04389762.

Keywords: Lactobacillus plantarum; PS128; Parkinson's disease; probiotics; psychobiotics.

Conflict of interest statement

Y-CT owns stock of Bened Biomedical Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Lu, Chang, Weng, Chen, Kuo and Tsai.

Figures

Figure 1
Figure 1
Study flow diagram.
Figure 2
Figure 2
The effect of PS128 intervention on metabolic parameters of (A) hs-CRP, (B) MPO, (C) TAC, (D) GPx, (E) 8-OHdG, and (F) CRE. *p < 0.05; **p < 0.01 as compared to baseline (V0). Mean ± SD; P, Obtained from paired t test; hs-CRP, high-sensitivity C-reactive protein; MPO, myeloperoxidase; TAC, total antioxidant capacity; GPx, glutathione peroxidase; 8-OHdG, 8-hydroxy-2'-deoxyguanosine; CRE, creatinine.

References

    1. Elfil M, Kamel S, Kandil M, Koo BB, Schaefer SM. Implications of the gut microbiome in Parkinson's disease. Mov Disord. (2020) 35:921–33. 10.1002/mds.28004
    1. Tysnes OB, Storstein A. Epidemiology of Parkinson's disease. J Neural Transm. (2017) 124:901–5. 10.1007/s00702-017-1686-y
    1. Tran TN, Vo TNN, Frei K, Truong DD. Levodopa-induced dyskinesia: clinical features, incidence, and risk factors. J Neural Transm. (2018) 125:1109–17. 10.1007/s00702-018-1900-6
    1. Zhang G, Xia Y, Wan F, Ma K, Guo X, Kou L, et al. . New perspectives on roles of alpha-synuclein in Parkinson's disease. Front Aging Neurosci. (2018) 10:370. 10.3389/fnagi.2018.00370
    1. Borghammer P, Van Den Berge N. Brain-first versus gut-first parkinson's disease: a hypothesis. J Parkinsons Dis. (2019) 9:S281–95. 10.3233/JPD-191721
    1. Liddle RA. Parkinson's disease from the gut. Brain Res. (2018) 1693(Pt B):201–6. 10.1016/j.brainres.2018.01.010
    1. Chandra R, Hiniker A, Kuo YM, Nussbaum RL, Liddle RA. alpha-Synuclein in gut endocrine cells and its implications for Parkinson's disease. JCI Insight. (2017) 2:e92295. 10.1172/jci.insight.92295
    1. Liu B, Fang F, Pedersen NL, Tillander A, Ludvigsson JF, Ekbom A, et al. . Vagotomy and Parkinson disease: a Swedish register-based matched-cohort study. Neurology. (2017) 88:1996–2002. 10.1212/WNL.0000000000003961
    1. Haikal C, Chen QQ, Li JY. Microbiome changes: an indicator of Parkinson's disease? Transl Neurodegener. (2019) 8:38. 10.1186/s40035-019-0175-7
    1. Gazerani P. Probiotics for Parkinson's disease. Int J Mol Sci. (2019) 20:4121. 10.3390/ijms20174121
    1. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. . Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. (2014) 11:506–14. 10.1038/nrgastro.2014.66
    1. Long-Smith C, O'Riordan KJ, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota-gut-brain axis: new therapeutic opportunities. Annu Rev Pharmacol Toxicol. (2020) 60:477–502. 10.1146/annurev-pharmtox-010919-023628
    1. Dinan TG, Stanton C, Cryan JF. Psychobiotics: a novel class of psychotropic. Biol Psychiatry. (2013) 74:720–6. 10.1016/j.biopsych.2013.05.001
    1. Liu WH, Chuang HL, Huang YT, Wu CC, Chou GT, Wang S, et al. . Alteration of behavior and monoamine levels attributable to Lactobacillus plantarum PS128 in germ-free mice. Behav Brain Res. (2016) 298(Pt B):202–9. 10.1016/j.bbr.2015.10.046
    1. Liu YW, Liu WH, Wu CC, Juan YC, Wu YC, Tsai HP, et al. . Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naive adult mice. Brain Res. (2016) 1631:1–12. 10.1016/j.brainres.2015.11.018
    1. Liao JF, Cheng YF, Li SW, Lee WT, Hsu CC, Wu CC, et al. . Lactobacillus plantarum PS128 ameliorates 2,5-Dimethoxy-4-iodoamphetamine-induced tic-like behaviors via its influences on the microbiota-gut-brain-axis. Brain Res Bull. (2019) 153:59–73. 10.1016/j.brainresbull.2019.07.027
    1. Bertaccini G. Tissue 5-hydroxytryptamine and urinary 5-hydroxyindoleacetic acid after partial or total removal of the gastro-intestinal tract in the rat. J Physiol. (1960) 153:239–49. 10.1113/jphysiol.1960.sp006532
    1. Liao JF, Cheng YF, You ST, Kuo WC, Huang CW, Chiou JJ, et al. . Lactobacillus plantarum PS128 alleviates neurodegenerative progression in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse models of Parkinson's disease. Brain Behav Immun. (2020) 90:26–46. 10.1016/j.bbi.2020.07.036
    1. Liu YW, Liong MT, Chung YE, Huang HY, Peng WS, Cheng YF, et al. . Effects of Lactobacillus plantarum PS128 on children with Autism Spectrum Disorder in Taiwan: a randomized, double-blind, placebo-controlled trial. Nutrients. (2019) 11:820. 10.3390/nu11040820
    1. Huang WC, Wei CC, Huang CC, Chen WL, Huang HY. The beneficial effects of Lactobacillus plantarum PS128 on high-intensity, exercise-induced oxidative stress, inflammation, and performance in Triathletes. Nutrients. (2019) 11:353. 10.3390/nu11020353
    1. Huang WC, Pan CH, Wei CC, Huang HY. Lactobacillus plantarum PS128 Improves physiological adaptation and performance in triathletes through gut microbiota modulation. Nutrients. (2020) 12:2315. 10.3390/nu12082315
    1. Liao PL, Wu CC, Chen TY, Tsai YC, Peng WS, Yang DJ, et al. . Toxicity studies of Lactobacillus plantarum PS128TM isolated from spontaneously fermented mustard greens. Foods. (2019) 8:668. 10.3390/foods8120668
    1. Otsuji S, Shibata H, Umeda M. Turbidimetric immunoassay of serum C-reactive protein. Clin Chem. (1982) 28:2121–4. 10.1093/clinchem/28.10.2121
    1. Benzie IF, Strain JJ. The ferric reducing ability of plasma (FRAP) as a measure of “antioxidant power”: the FRAP assay. Anal Biochem. (1996) 239:70–6. 10.1006/abio.1996.0292
    1. Marsden CD. Slowness of movement in Parkinson's disease. Mov Disord. (1989) 4(Suppl 1):S26–37. 10.1002/mds.870040505
    1. Hallett M. Bradykinesia: why do Parkinson's patients have it and what trouble does it cause? Mov Disord. (2011) 26:1579–81. 10.1002/mds.23730
    1. Tamtaji OR, Taghizadeh M, Daneshvar Kakhaki R, Kouchaki E, Bahmani F, Borzabadi S. Clinical and metabolic response to probiotic administration in people with Parkinson's disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr. (2019) 38:1031–5. 10.1016/j.clnu.2018.05.018
    1. Borzabadi S, Oryan S, Eidi A, Aghadavod E, Daneshvar Kakhaki R, Tamtaji OR. The effects of probiotic supplementation on gene expression related to inflammation, insulin and lipid in patients with Parkinson's disease: a randomized, double-blind, placebocontrolled trial. Arch Iran Med. (2018) 21:289–95.
    1. Georgescu D, Ancusa OE, Georgescu LA, Ionita I, Reisz D. Nonmotor gastrointestinal disorders in older patients with Parkinson's disease: is there hope? Clin Interv Aging. (2016) 11:1601–8. 10.2147/CIA.S106284
    1. Barichella M, Pacchetti C, Bolliri C, Cassani E, Iorio L, Pusani C, et al. . Probiotics and prebiotic fiber for constipation associated with Parkinson disease: an RCT. Neurology. (2016) 87:1274–80. 10.1212/WNL.0000000000003127
    1. Cassani E, Privitera G, Pezzoli G, Pusani C, Madio C, Iorio L, et al. . Use of probiotics for the treatment of constipation in Parkinson's disease patients. Minerva Gastroenterol Dietol. (2011) 57:117–21.
    1. Malle E, Buch T, Grone HJ. Myeloperoxidase in kidney disease. Kidney Int. (2003) 64:1956–67. 10.1046/j.1523-1755.2003.00336.x
    1. Choi DK, Pennathur S, Perier C, Tieu K, Teismann P, Wu DC, et al. . Ablation of the inflammatory enzyme myeloperoxidase mitigates features of Parkinson's disease in mice. J Neurosci. (2005) 25:6594–600. 10.1523/JNEUROSCI.0970-05.2005
    1. Maki RA, Holzer M, Motamedchaboki K, Malle E, Masliah E, Marsche G, et al. . Human myeloperoxidase (hMPO) is expressed in neurons in the substantia nigra in Parkinson's disease and in the hMPO-alpha-synuclein-A53T mouse model, correlating with increased nitration and aggregation of alpha-synuclein and exacerbation of motor impairment. Free Radic Biol Med. (2019) 141:115–40. 10.1016/j.freeradbiomed.2019.05.033
    1. Jucaite A, Svenningsson P, Rinne JO, Cselenyi Z, Varnas K, Johnstrom P, et al. . Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson's disease. Brain. (2015) 138(Pt 9):2687–700. 10.1093/brain/awv184
    1. Zhang H, Ray A, Miller NM, Hartwig D, Pritchard KA, Dittel BN. Inhibition of myeloperoxidase at the peak of experimental autoimmune encephalomyelitis restores blood-brain barrier integrity and ameliorates disease severity. J Neurochem. (2016) 136:826–36. 10.1111/jnc.13426
    1. Stefanova N, Georgievska B, Eriksson H, Poewe W, Wenning GK. Myeloperoxidase inhibition ameliorates multiple system atrophy-like degeneration in a transgenic mouse model. Neurotox Res. (2012) 21:393–404. 10.1007/s12640-011-9294-3
    1. Kaindlstorfer C, Sommer P, Georgievska B, Mather RJ, Kugler AR, Poewe W, et al. . Failure of neuroprotection despite microglial suppression by delayed-start myeloperoxidase inhibition in a model of advanced multiple system atrophy: clinical implications. Neurotox Res. (2015) 28:185–94. 10.1007/s12640-015-9547-7
    1. Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Inflammation and adaptive immunity in Parkinson's disease. Cold Spring Harb Perspect Med. (2012) 2:a009381. 10.1101/cshperspect.a009381
    1. Mirza B, Hadberg H, Thomsen P, Moos T. The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease. Neuroscience. (2000) 95:425–32. 10.1016/S0306-4522(99)00455-8
    1. Blum-Degen D, Muller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neurosci Lett. (1995) 202:17–20. 10.1016/0304-3940(95)12192-7
    1. Mogi M, Harada M, Narabayashi H, Inagaki H, Minami M, Nagatsu T. Interleukin (IL)-1 beta, IL-2, IL-4, IL-6 and transforming growth factor-alpha levels are elevated in ventricular cerebrospinal fluid in juvenile parkinsonism and Parkinson's disease. Neurosci Lett. (1996) 211:13–6. 10.1016/0304-3940(96)12706-3
    1. Pfeiffer RF. Gastrointestinal dysfunction in Parkinson's disease. Lancet Neurol. (2003) 2:107–16. 10.1016/S1474-4422(03)00307-7
    1. Barone P, Antonini A, Colosimo C, Marconi R, Morgante L, Avarello TP, et al. . The PRIAMO study: a multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson's disease. Mov Disord. (2009) 24:1641–9. 10.1002/mds.22643
    1. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. (2003) 24:197–211. 10.1016/S0197-4580(02)00065-9
    1. Braak H, Del Tredici K. Neuroanatomy and pathology of sporadic Parkinson's disease. Adv Anat Embryol Cell Biol. (2009) 201:1–119.
    1. Corbille AG, Letournel F, Kordower JH, Lee J, Shanes E, Neunlist M, et al. . Evaluation of alpha-synuclein immunohistochemical methods for the detection of Lewy-type synucleinopathy in gastrointestinal biopsies. Acta Neuropathol Commun. (2016) 4:35. 10.1186/s40478-016-0305-8
    1. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, et al. . Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models parkinson's disease. Neuron. (2019) 103:627–41.e7. 10.1016/j.neuron.2019.05.035
    1. Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. . The microbiota-gut-brain axis. Physiol Rev. (2019) 99:1877–2013. 10.1152/physrev.00018.2018
    1. Cryan JF, O'Riordan KJ, Sandhu K, Peterson V, Dinan TG. The gut microbiome in neurological disorders. Lancet Neurol. (2020) 19:179–94. 10.1016/S1474-4422(19)30356-4
    1. Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, et al. . Colonic bacterial composition in Parkinson's disease. Mov Disord. (2015) 30:1351–60. 10.1002/mds.26307
    1. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, et al. . Gut microbiota are related to Parkinson's disease and clinical phenotype. Mov Disord. (2015) 30:350–8. 10.1002/mds.26069
    1. Heintz-Buschart A, Pandey U, Wicke T, Sixel-Doring F, Janzen A, Sittig-Wiegand E, et al. . The nasal and gut microbiome in Parkinson's disease and idiopathic rapid eye movement sleep behavior disorder. Mov Disord. (2018) 33:88–98. 10.1002/mds.27105

Source: PubMed

3
Iratkozz fel